1,632
Views
0
CrossRef citations to date
0
Altmetric
Novel Vaccines

A systematic review on malaria and dengue vaccines for the effective management of these mosquito borne diseases: Improving public health

, , ORCID Icon, , ORCID Icon, ORCID Icon, , ORCID Icon, , ORCID Icon, , & show all
Article: 2337985 | Received 17 Feb 2024, Accepted 28 Mar 2024, Published online: 11 Apr 2024

ABSTRACT

Insect vector-borne diseases (VBDs) pose significant global health challenges, particularly in tropical and subtropical regions. The WHO has launched the “Global Vector Control Response (GVCR) 2017–2030” to address these diseases, emphasizing a comprehensive approach to vector control. This systematic review investigates the potential of malaria and dengue vaccines in controlling mosquito-borne VBDs, aiming to alleviate disease burdens and enhance public health. Following PRISMA 2020 guidelines, the review incorporated 39 new studies out of 934 identified records. It encompasses various studies assessing malaria and dengue vaccines, emphasizing the significance of vaccination as a preventive measure. The findings indicate variations in vaccine efficacy, duration of protection, and safety considerations for each disease, influencing public health strategies. The review underscores the urgent need for vaccines to combat the increasing burden of VBDs like malaria and dengue, advocating for ongoing research and investment in vaccine development.

Introduction

Vector-borne diseases (VBD) represent a significant public health burden globally, causing substantial morbidity and mortality in human populations.Citation1–3 These diseases, encompassing well-known illnesses like malaria, dengue fever, chikungunya, Zika virus, yellow fever, and Lyme disease, are primarily transmitted through the bite of infected arthropod vectors such as mosquitoes, ticks, and fleas.Citation4 Whereas, diseases like Chagas and epidemic typhus can be transmitted through other means, such as blood transfusion or fecal contamination.Citation1,Citation5 These diseases, caused by parasites, bacteria, or viruses, account for over 700,000 annual deaths, constituting more than 17% of infectious disease-related fatalities.Citation6 Moreover, there is a threat to more than 80% population of the world, disproportionately affecting the poorest populations living in the tropics and subtropics.Citation7 Globally, the complex triad of vector-pathogen-host interactions in the transmission cycle makes the prevention and control of VBD a complex challenge for healthcare systems.Citation8 Malaria, one of the most prevalent vector-borne diseases, is transmitted through Anopheline mosquitoes and results in approximately 219 million cases and over 400,000 deaths each year, with children under 5 years old bearing the brunt of this burden.Citation9,Citation10 Similarly, dengue, primarily carried by Aedes mosquitoes, threatens over 3.9 billion people in 129 countries, resulting in an estimated 96 million symptomatic cases and around 40,000 deaths yearly.Citation11,Citation12 The impact of these diseases is especially pronounced in tropical and subtropical regions, disproportionately affecting the most disadvantaged populations.Citation10 Nevertheless, several outbreaks of these diseases have surged among developed countries since 2014 owing to globalization, urbanization and drastic climate changes, straining healthcare systems and claiming numerous lives.Citation13,Citation14

To combat these diseases, the World Health Organization (WHO) has developed the “Global Vector Control Response (GVCR) 2017–2030,” which offers guidance to nations and partners to strengthen vector control for disease prevention and outbreak response.Citation15 This initiative calls for program realignment, technical capacity enhancement, improved infrastructure, enhanced monitoring, and community mobilization. By taking a comprehensive approach to vector control, it aligns with national and global health goals, including Sustainable Development Goals (SDGs) and Universal Health Coverage.Citation16 Traditionally, vector control measures have primarily relied on insecticides, environmental management, and public health campaigns to reduce vector populations and prevent disease transmission.Citation17 While insecticide-treated bednets and indoor residual spraying have undoubtedly demonstrated success; however, their sustained efficacy is threatened by the emergence of resistance, necessitating a comprehensive approach that includes innovative strategies, such as vaccine development, to counteract the evolving landscape of vector-borne diseases.Citation18 The development of vaccines for malaria and dengue has been a focus of intense research efforts due to the significant global health impact of these diseases.Citation19 Vaccine research for Plasmodium parasites has been a central focus for almost six decades, the integration of effective vaccines into malaria control programs would signify a pivotal and revolutionary paradigm shift in combatting VBDs, offering a complementary strategy to established interventions like insecticide-treated bednets and indoor residual spraying.Citation20 The development of the RTS,S/AS01 vaccine, marketed as Mosquirix, represents a significant breakthrough.Citation21 It is the first, and currently the only, vaccine licensed for use against a parasitic disease in humans. The vaccine has shown partial protection against malaria in children in sub-Saharan Africa, highlighting a critical step forward in the fight against this disease.Citation22 Dengue vaccine development has also seen significant progress, notably with the creation and licensing of Dengvaxia (CYD-TDV) by Sanofi Pasteur.Citation23 This vaccine is recommended for individuals 9–45 years old living in dengue-endemic areas and has been approved in several countries. However, its deployment is complex due to its varying efficacy against the four different dengue virus serotypes and the need to pre-screen for prior dengue exposure to avoid the risk of severe dengue in those not previously infected.Citation24 By harnessing the potential of vaccines in the management of VBD, there is substantial potential for making significant strides in reducing the disease burden and improving the well-being of affected populations. This systematic review seeks to provide a comprehensive and critical analysis of the current status, challenges, and opportunities in utilizing vaccination as a key strategy for managing mosquito VBD particularly focusing on malaria and dengue, ultimately contributing to the advancement of public health initiatives worldwide. Subsequently this review aims to identify key research gaps and offer insights into future directions for vaccine development, deployment, and implementation strategies to address the challenges in combating insect VBD.

Materials and methods

Study protocol and registration

This systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guidelines.Citation25 A comprehensive review protocol was developed before initiating the review process. The protocol outlined the review’s objectives, inclusion and exclusion criteria, search strategy, data extraction methods, and the plan for assessing the quality of included studies.

Eligibility criteria

Population

Studies involving humans, both adults and children, residing in regions affected by insect VBDs. There were no restrictions based on age, gender, or comorbidities.

Intervention

Studies evaluating vaccines designed for the prevention or management of insect vector-borne diseases, including dengue fever and malaria. Both traditional and novel vaccine candidates were considered.

Comparators

Studies comparing vaccinated individuals with unvaccinated or differently vaccinated individuals were included. In cases where no comparator group was available, observational studies assessing vaccine effectiveness were considered.

Outcomes

The primary outcomes of interest included vaccine safety, immunogenicity, efficacy, and effectiveness. Secondary outcomes encompassed adverse events, duration of protection, and potential complications associated with vaccines. Additionally, studies reporting on the implications of vaccine use, including public health impact, ethical considerations, and logistical challenges, were included.

Inclusion and exclusion criteria

Inclusion criteria encompassed peer-reviewed articles and clinical trials published in English, focusing on vaccines or vaccination strategies for malaria or dengue. The selected studies comprised studies emphasizing outcomes related to vaccine efficacy, safety, immunogenicity, and adverse events. Conversely, exclusion criteria involved non-peer-reviewed articles, editorials, letters, and conference abstracts, along with studies in languages other than English, those unrelated to vaccines or vaccination strategies for the specified diseases, animal studies, in vitro studies, and modeling studies lacking empirical data. Studies devoid of relevant data on vaccine efficacy, safety, or immunogenicity were also excluded.

Study designs

Randomized controlled trials (RCTs) and observational studies (cohort studies, case-control studies) were considered for inclusion.

Search strategy

A comprehensive search strategy was developed to identify relevant studies. The following electronic databases were systematically searched from inception to August 2023: PubMed, Embase, Google Scholar, Web of Science, and the Cochrane Library. The search strategy combined Medical Subject Headings (MeSH) terms, keywords, and tailored to each database’s syntax. The search strategy was piloted and refined iteratively to ensure sensitivity and comprehensiveness. The search terms employed a combination of keywords related to insect vector-borne diseases (VBDs), vaccination, and disease management. The search string included but was not limited to the following keywords: “vector-borne diseases,” “mosquito-borne diseases,” “malaria,” “dengue,” “vaccine,” “vaccination,” “immunization,” “preventive measures,” and “public health.” Boolean operators (AND, OR) were employed to refine the search and capture relevant articles pertaining to the intersection of vaccines and insect VBD.

Study selection

Two independent reviewers conducted the initial title and abstract screening using predefined inclusion and exclusion criteria. Full-text articles of potentially eligible studies were then assessed independently by the same reviewers. Discrepancies were resolved through discussion, and a third reviewer was consulted if consensus could not be reached.

Data extraction

A standardized data extraction form was developed and used to collect relevant information from the included studies. Data extraction included study characteristics (e.g., author, publication year, country, study design, study duration), participant demographics (vector-borne disease, sample size for both vaccinated and control/placebo group), vaccine details, outcomes of interest, and any additional relevant information (follow-up and findings). Data extraction was performed independently by two reviewers, with a third reviewer available to resolve discrepancies. A narrative synthesis of the findings from the included studies was performed, structured around the outcomes of interest. Qualitative synthesis was supplemented by tabulation of relevant study characteristics, vaccine details, and outcomes.

Quality assessment

The quality and risk of bias assessment for included studies were conducted using appropriate tools tailored to the study design. For RCTs, the Cochrane Risk of Bias tool 2.0 was used, while the Newcastle-Ottawa Scale (NOS) was employed for observational studies. The GRADE approach was utilized to assess the overall quality of evidence for each outcome. Visualization of Risk of Bias assessment is generated by Robvis Tool.

Ethical considerations

Ethical approval was not required for this systematic review as it involved the analysis of publicly available data from previously published studies.

Results

In this systematic review, the PRISMA flow diagram illustrates the comprehensive process of study selection. Initially, 934 records were identified from various databases. After eliminating 411 duplicate records, the researchers screened 523 unique records. During the screening phase, 249 records were excluded based on criteria like not pertaining to VBD vaccines. This left the researchers with 274 records for which they sought full-text articles. Following a thorough assessment of these articles for eligibility, 107 reports were excluded due to factors such as being unrelated to VBD vaccines, not in English, or other reasons. Ultimately, the systematic review included 39 new studies, reflecting a meticulous and rigorous process in the selection of relevant research for the review ().

Figure 1. Prisma flow diagram.

Figure 1. Prisma flow diagram.

Dengue vaccines

While the studies in collectively highlight the potential of Dengue vaccines, they vary in terms of study designs, target populations, vaccine types, and follow-up durations. Some studies, like those by Rivera et al. and Kallas et al., focus on vaccine efficacy over several years. Others, such as Sridhar et al. and Vannice et al., emphasize the protection of individuals with prior Dengue exposure. The studies provide a valuable foundation for understanding the safety, immunogenicity, and efficacy of Dengue vaccines across diverse populations and regions. Notably, CYD-TDV demonstrated good tolerability over three years in Dengue-endemic regions among individuals aged 1.5–45 years. TAK-003 displayed efficacy against symptomatic Dengue in pediatric populations across various countries over three years. Dengvaxia post-licensure studies, spanning four years among pediatric populations, emphasized robust surveillance and risk management. Butantan-DV and TV003 vaccines induced balanced neutralizing antibody responses across all four DENV serotypes in adults, whether naïve or exposed. CYD-TDV exhibited efficacy with a satisfactory safety profile, meeting WHO standards but maintained Dengue mortality rates at 50%. These diverse results underscore promising avenues for Dengue vaccine development but highlight the need for enhanced efficacy and mortality rate reduction. Moreover, the TV003 trial in the USA observed a significant antibody response in 90% of flavivirus-naive subjects within a six-month follow-up.

Table 1. Key attributes of published research on dengue vaccination.

Malaria vaccines

The presents a compilation of studies on malaria vaccines conducted across various countries and populations. These studies collectively demonstrate significant progress in the development of malaria vaccines, with varying levels of efficacy, safety, and immunogenicity. RTS, S/AS01 exhibits promise in safeguarding against clinical and severe malaria, with efficacy spanning 12 to 19 months. Conversely, GMZ2 displays limited efficacy despite reducing malaria incidence within a six-month follow-up. R21/Matrix-M booster doses maintain high efficacy against multiple episodes of clinical malaria. R21/MM, while safe and immunogenic, demonstrates high-level efficacy for 12 months. RTS, S/AS01‘s greater activity against malaria parasites in children aged 5–17 months is evident in Kenya and across seven countries. Additionally, in different African sites, RTS, S/AS01 showcases potential in combination with other control measures. DNA/MVA heterologous prime-boost vaccination proves safe and highly immunogenic for effector T cell induction. Notably, two RTS, S/AS01E formulations exhibit equal safety and immunogenicity, providing modest protection against malaria when co-administered with EPI vaccines.

Table 2. Key attributes of published research on malaria vaccination.

Risk of assessment

The quality assessment of the included studies reveals variations in the risk of bias across different domains as described in and . Risk of assessment was carried out for the randomized controlled trials (RCTs) in this systematic review to validate the results and ensure that the design, conduct, and reporting of RCTs are of high quality, helping the reviewers and readers to determine the degree to which the study results can be attributed to the intervention rather than methodological flaws as well as minimizing the systematic errors as it is crucial for drawing accurate conclusion about the effectiveness of interventions.Citation66 A rigorous risk of bias assessment in this systematic review of RCTs was carried out to ensure the reliability and acceptability of the synthesized evidence putting a step forward in contributing to evidence based decision-making in healthcare, informing both clinical practice and future research endeavors. Cochrane Collaboration introduced a risk of bias tool to assess the internal validity of randomized controlled trials in February 2008. The risk of bias tool comprises six domains: sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and “other sources of bias.” Distinct evaluations of the risk of bias (categorized as high, low, or unclear) are conducted for each individual domain. The ultimate comprehensive evaluation, either within a single study or across multiple studies, is determined by the collective assessments of individual domains.Citation67

Figure 2. Visualization of risk of bias assessment is generated by robvis tool.Citation65

Figure 2. Visualization of risk of bias assessment is generated by robvis tool.Citation65

Table 3. Risk of bias assessment for randomized controlled trials.

represents the traffic light visualization of risk of bias assessment of RCTs in this systematic review. This color coded system is user friendly, allowing researchers, clinicians and readers to quickly gasp the overall risk of bias in different domains of a study. Green color indicates Low risk, suggesting high level of methodological rigor and reliability. Yellow color signals an unclear risk of bias i.e., there might be some concerns or uncertainties regarding the evaluation of risk of bias in selected domain as the information provided in those RCTs is insufficient to clearly categorize it either low or high risk. Red color signifies high risk of bias depicting a potential impact on the validity of the study results. This visualization is generated by Robvis Tool (a web app designed for visualizing risk of bias assessments performed as a part of systematic review). Quality assessment of cohort studies was done using Newcastle-Ottawa scale (NOS) to ensure the reliability and validity of results. Higher quality score indicates that it has met the quality standards.

Discussion

In this systematic review, our exploration into the realm of insect VBDs and their management through vaccination revealed significant insights into the efficacy, challenges, and future prospects of vaccination strategies. Our synthesis of the literature highlighted several key findings: the efficacy of existing vaccines, challenges in deployment and accessibility, the role of adjuvants, and promising avenues for future research and vaccine development. This discussion aims to delve deeper into these key facets, shedding light on the implications of these findings in the broader context of public health and disease management.

Dengue poses a significant public health threat, with potentially severe outcomes, including hospitalizations and mortality. As such, the development of effective vaccines has been a critical endeavor in reducing the burden of this disease.Citation68 Studies are being carried out in the context of the global Strategy for Dengue Prevention and Control, 2012–2020 by WHO which aims to address this need and at least reduce the mortality and morbidity rates by 50% and 25% respectively by the end of 2020.Citation39,Citation69 A critical literature review shows that the dengue vaccine TAK-003 demonstrated a high efficacy over 3 years against symptomatic dengue. A clinical trial in the pediatric population, conducted across 8 dengue-endemic countries showed that Takeda’s dengue vaccine is efficacious against symptomatic dengue over 3 years. The efficacy may decline over time but its robustness remained intact in hospitalized dengue.Citation26,Citation70 Among the 4 different serotypes of dengue, TAK-003 shows a high level of dengue serotype 2 neutralizing antibodies while the other serotypes 1, 3, and 4 responses are lower.Citation71 TAK-003 has a sustained long-term efficacy overall dengue in hospitalized individuals without any increased risk of hospitalization and important safety risks.Citation72 Butantan-DV and TV003 are analogous to each other and effective and well-tolerated against DENV-naive and DENV-exposed participants. Seroconversion is achieved for all DENV serotypes.Citation27,Citation73 The rash is a common systemic adverse effect and it is associated with a tetravalent immune response.Citation74 The immunogenicity of Butantan-DV is elucidated by its plasmablast expansion in the blood and its ability to induce B-cell activation.Citation75 Dengvaxia hereafter referred to as CYP-TDV is a live, attenuated, recombinant, tetravalent dengue vaccine that is the first vaccine approved for the prevention of symptomatic dengue in individuals aged 9–60 years by any of the 4 DENV serotypes in many endemic areas.Citation31,Citation76-78 Studies are conducted for the safety and efficacy of CYD-TDV after thoroughly assessing the potential risks and a risk-minimization action plan was defined subsequent review of data generated thereafter was followed by the World Health Organization (WHO), Pan American Health Organization, Centers for Disease Control and Prevention, key opinion leaders, and regulatory agencies.Citation31 A phase 3 clinical trial conducted in Latin America shows the desired efficacy of CYD-TDV in symptomatic VCD, hospitalization for dengue, and severe dengue in children aged 9–16 years after three-dose vaccination scheduleCitation32 that lines with findings of similarly designed Asian trials.Citation33 In a Phase III trial of CYD-TDV efficacy after 1 or 2 doses in dengue endemic areas showed that CYD-TDV efficacy was null to modest in the seronegative participants of any age group after any dose.Citation79 The decreased effectiveness of the CYD-TDV vaccine against DENV2, a notably severe strain associated with dengue outbreaks, raises significant concerns about its ability to provide adequate protection.Citation80 The risk of severe dengue (DHF/DSS) is recognized to be greater during a secondary infection compared to primary infection,Citation81,Citation82 thus there is a theoretical proposition suggesting that administering CYD-TDV vaccination to an individual lacking prior dengue exposure may mimic a primary infection, potentially heightening the susceptibility to severe dengue following a subsequent natural infection post-vaccination.Citation83

Phase II clinical studies in Singapore and Vietnam showed persistent anti-dengue antibodies over 5 years with no safety concerns and suggested the use of a three-dose schedule at 0-, 6-, and 12 months in those with prior dengue exposure.Citation84 Ongoing safety and efficacy assessment of live attenuated vaccine by manufacturer’s risk management plan (RMP) and post-licensure monitoring played an integral role and the countries that introduce the vaccine are encouraged to conduct their post-licensure monitoring and evaluation which in turn requires planning and strengthening of vaccine surveillance.Citation29 Currently, CYD-TDV is licensed and available in 20 countries for the population age 9–45 years old. According to Sanofi Pasteur’s official guidelines, the World Health Organization (WHO) recommends the administration of CYD-TDV specifically to individuals with documented prior dengue virus infections. This strategic approach aims to mitigate the potential risk of severe dengue should these individuals encounter the virus after vaccination. The data analyzed from three efficacy trials elucidated this concern and showed that CYD-TDV has a protective effect for 5 years in seropositive persons against VCD and hospitalization for VCD but there is evidence of a higher risk of VCD and hospitalization for VCD in persons who are not exposed to dengue.Citation28

Malaria stands as a prominent global health burden within the spectrum of VBDs, exerting a substantial impact on public health by contributing significantly to both morbidity and mortality. It is estimated that nearly half of the world’s population resides in regions where the risk of malaria transmission prevails.Citation16 In the World Malaria Report 2019 issued by the WHO, it was reported that in 2018, there were an estimated 228 million cases of malaria and 405,000 malaria-induced fatalities documented on a worldwide scale. Malaria can be attributed to any of the 8 Plasmodium species, with the majority of malaria cases being attributed to P. falciparum and P. vivax. However, fatalities primarily result from falciparum malaria.Citation85–87 Malaria is endemic in over 90 countries, impacting an estimated 40% of the global population.Citation88 A recombinant protein malaria vaccine GMZ2 is prepared, containing the conserved fragments of two blood-stage antigens of Plasmodium falciparum, glutamate-rich protein (GLURP) and merozoite surface protein 3 (MSP3)Citation89 both of these proteins have been recognized as subjects of naturally acquired immunity to malariaCitation90,Citation91 and stimulates the production of specific and functional antibodies capable of controlling parasite replication at high levels.Citation92,Citation93 GMZ2 demonstrated good tolerability and immunogenicity, leading to a reduction in malaria incidence.Citation93–95 However, to fulfill a potential public health role, the vaccine’s efficacy would require significant improvement through the development of a more immunogenic formulation.Citation44

The development of an efficacious malaria vaccine has posed a formidable challenge within the realm of medical science. Nevertheless, remarkable progress has been achieved on a global scale in the endeavor to combat malaria. In light of this concerning trend necessitating the emergence of innovative tools to address the disease, the RTS, S vaccine has been introduced at a pivotal moment. In 1987 by the collaboration of GlaxoSmithKline (GSK) and the Walter Reed Army Institute of Research (WRAIR), the RTS, S vaccine was created.Citation96 In the journey of testing multiple adjuvants of this vaccine AS01 adjuvant system which comprises liposomes MPL, and QS-21, provided an opportunity to improve RTS, S immunogenicity.Citation97–99 Comparative field trials between RTS, S/AS01B, and RTS, S/AS02A formulations demonstrated a more advantageous immunogenicity profile of RTS, S/AS01B as compared to RTS, S/AS02A while both vaccines have favorable tolerability during a 12-month surveillance period.Citation100,Citation101 The phase 3 trial of the RTS, S/AS01 malaria vaccine elicited a safe and efficacious response against clinical and severe malaria in all age groups, reducing the burden of overall disease and improving the health outcomes in the areas endemic to malaria caused by P. falciparum.Citation46Citation47-49Citation54-102Citation103 On October 6, 2021, WHO approved RTS, S AS01 for widespread use. The vaccine demonstrates a significant reduction in both the overall incidence of malaria cases and the incidence of severe, life-threatening malaria among young children.

Plasmodium falciparum circumsporozoite (CS) and hepatitis B surface (HBs) antigens when assessed and cell-mediated immune responses, the anti-CS antibody response was predominantly higher with RTS, S/AS01 than with RTS, S/AS02,Citation101Citation104–109 notably, both vaccines have acceptable safety profile and higher reactogenicity as compared to the non-adjuvanted RTS, S.Citation110,Citation111 RTS, S/AS01 induced strong humoral immunity and cell-mediated immunity in all pre-clinical and clinical trials when tested with various antigens and it has been considered safe to use in adults as well as pediatrics.Citation57 In the context of vaccine adjuvants, it’s important to note that inflammation plays a pivotal role in initiating immune responses. This inflammation can result from the activation of specific receptors or nonspecific activation, and it represents the primary mechanism by which adjuvants stimulate antigen-specific cellular immunity.Citation16

A next-generation RTS, S-like vaccine/improved version of RTS, S, the R21 was developed by Jenner Institute in Oxford, UK. After conducting preclinical studies with R21 plus various adjuvants, Matrix-M (R21/MM) was chosen for clinical development due to its notable immunogenicity.Citation112 In phase 1/2a clinical trials, R21/MM showed a favorable safety profile and strong antibody responses.Citation43,Citation43,Citation113 In Nanoro, Burkina Faso, one of the 11 trial sites for the RTS, S/AS01 phase 3 trial, the R21 vaccine combined with a higher dose of adjuvant MM demonstrated a superior efficacy of 77%. This outperformed the 44% efficacy observed with RTS, S/AS01 at the same site over a 12-month follow-up, without any planned seasonal administration.Citation114 The adverse events profile of the R21 vaccine shows fewer events than RTS, S AS01.Citation43 Over 2 years of follow-up after the primary series of vaccination, high efficacy of R21/Matrix-M malaria vaccine is reported which reached the WHO efficacy goal of 75% or greater in the target population of African children over 24 months. The R21/Matrix-M vaccine maintains a satisfactory safety profile during the second year of follow-up, even after the administration of a fourth dose.Citation42 Following RTS, S AS01, R21/Matrix-M vaccine is the second malaria vaccine that is recommended by WHO for malaria prevention in its updated advice on immunization published on October 2, 2023, due to its high efficacy when administered before the high transmission season, good efficacy in an aged-based schedule, high impact, cost-effectiveness similarity with RTS, S vaccine and safety.Citation115

Viral vectors hold substantial promise in vaccine development as they facilitate intracellular antigen expression, enhancing the capacity to elicit potent cytotoxic T-lymphocyte responses and pro-inflammatory interferon and cytokine production without any adjuvants.Citation116 Multiple epitopes (ME)-thrombospondin-related adhesion protein (TRAP) candidate vaccines against Plasmodium falciparum are engineered to effectively stimulate effector T cells.Citation48 In a Phase IIb trial conducted in malaria-endemic areas in Kenya, both ChAd63 ME-TRAP and MVA ME-TRAP vaccines demonstrated some protective efficacy.Citation117 The prime-boost vaccination regimen involving ChAd63 and MVA ME-TRAP has exhibited a satisfactory safety profile across four cohorts of children with progressively younger ages in The Gambia and Burkina Faso, MVA ME-TRAP being more reactogenic than ChAd63 and indicated greater AE incidence.Citation118 The immunogenicity is reduced in the participants who are previously exposed to malariaCitation119 thus in this context DNA/MVA heterologous prime-boost vaccination resulted in a significant decrease in liver-stage parasites during challenge studies involving nonimmune volunteers but this initial T cell-inducing vaccine demonstrated ineffectiveness in reducing the natural infection rate among semi-immune African adults.Citation48

While the systematic review on vaccines for the effective management of insect VBD provides valuable insights, it also has few limitations. The quality of the studies included in the review varies. Some studies may have a higher risk of bias, impacting the overall reliability of the findings. The review might also be subjected to publication bias including language bias, as it mainly relies on published literature, potentially excluding unpublished studies or reports, leading to an incomplete representation of available data. The review doesn’t account for the real-world availability and implementation of these vaccines, which can significantly impact their public health impact. The review covers studies conducted over various timeframes. Changes in disease prevalence, vector behavior, and healthcare practices over time may affect the relevance of the findings. VBD and the effectiveness of vaccines can vary by region. The review doesn’t always distinguish between these regional variations, potentially oversimplifying the findings. Some studies have relatively short follow-up periods, which might not capture the long-term safety and efficacy of vaccines. Nevertheless, our manuscript offers a consolidated and critical analysis that synthesizes current knowledge, identifies gaps, and emphasizes the importance of sustained efforts in vaccine development and deployment to combat insect VBD effectively. Globally, physicians and pharmacies play a pivotal role in educating, advocating, and providing access to vaccines against VBD. Their expertise, advocacy, and accessibility significantly impact the public’s perception, acceptance, and access to these preventive measures, ultimately contributing to effective disease management and public health improvement on a global scale.

Conclusions

In conclusion, VBDs, notably dengue and malaria, pose significant global health threats, especially in tropical regions. Vaccines like TAK-003 and Butantan-DV exhibit potential against dengue, with TAK-003 showing efficacy over three years and Butantan-DV effective against various DENV serotypes. GMZ2 demonstrates promise in reducing malaria incidence, while RTS, S/AS01 marks a milestone, providing protection against clinical and severe malaria, especially in children. R21/Matrix-M enhances effectiveness and safety, earning WHO recommendation. Viral vector-based vaccines show potential against Plasmodium falciparum, but challenges persist in heavily affected regions. The review consolidates advancements in dengue and malaria vaccines, highlighting TAK-003, Butantan-DV, RTS, S/AS01, and R21/Matrix-M. It underscores the importance of viral vector-based vaccines and the 17D-derived vaccine in preventing yellow fever outbreaks. Further research is crucial to tackle the complex challenges posed by these diseases globally.

Author contributions

All authors have adhered to the guidelines and participated in the creation of the manuscript. They have reviewed and approved the final version for publication.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Swei A, Couper LI, Coffey LL, Kapan D, Bennett S. Patterns, drivers, and challenges of vector-borne disease emergence. Vector-Borne Zoonotic Dis. 2020;20(3):159–16. doi:10.1089/vbz.2018.2432.
  • Petersen LR, Beard CB, Visser SN. Combatting the increasing threat of vector-borne disease in the United States with a national vector-borne disease prevention and control system. Am J Trop Med Hyg. 2019;100(2):242. doi:10.4269/ajtmh.18-0841.
  • Eder M, Cortes F, Teixeira de Siqueira Filha N, Araújo de França GV, Degroote S, Braga C, Ridde V, Turchi Martelli CM. Scoping review on vector-borne diseases in urban areas: transmission dynamics, vectorial capacity and co-infection. Infect Dis Poverty. 2018;7(1):1–24. doi:10.1186/s40249-018-0475-7.
  • Chala B, Hamde F. Emerging and re-emerging vector-borne infectious diseases and the challenges for control: a review. Front Public Health. 2021;9:715759. doi:10.3389/fpubh.2021.715759.
  • Chao C, Leone JL, Vigliano CA. Chagas disease: historic perspective. Biochim Biophys Acta (BBA)-Mol Basis Dis. 2020;1866(5):165689. doi:10.1016/j.bbadis.2020.165689.
  • Suk JE, Van Cangh T, Beaute J, Bartels C, Tsolova S, Pharris A, Ciotti M, Semenza JC. The interconnected and cross-border nature of risks posed by infectious diseases. Glob Health Action. 2014;7(1):25287. doi:10.3402/gha.v7.25287.
  • Golding N, Wilson AL, Moyes CL, Cano J, Pigott DM, Velayudhan R, Brooker SJ, Smith DL, Hay SI, Lindsay SW, et al. Integrating vector control across diseases. BMC Med. 2015;13(1):249. doi:10.1186/s12916-015-0491-4.
  • Wikel S. Ticks and tick-borne pathogens at the cutaneous interface: host defenses, tick countermeasures, and a suitable environment for pathogen establishment. Front Microbiol. 2013;4:337. doi:10.3389/fmicb.2013.00337.
  • Sachs J, Malaney P. The economic and social burden of malaria. Nature. 2002;415(6872):680–5. doi:10.1038/415680a.
  • O’Meara WP, Mangeni JN, Steketee R, Greenwood B. Changes in the burden of malaria in sub-Saharan Africa. Lancet Infect Dis. 2010;10(8):545–55. doi:10.1016/S1473-3099(10)70096-7.
  • Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM, Brownstein JS, Hoen AG, Sankoh O, et al. The global distribution and burden of dengue. Nature. 2013;496(7446):504–7. doi:10.1038/nature12060.
  • Shepard DS, Undurraga EA, Halasa YA. Economic and disease burden of dengue in Southeast Asia. PLOS Negl Trop Dis. 2013;7(2):e2055. doi:10.1371/journal.pntd.0002055.
  • Semenza JC, Suk JE. Vector-borne diseases and climate change: a European perspective. FEMS Microbiol Lett. 2018;365(2):fnx244. doi:10.1093/femsle/fnx244.
  • Medlock JM, Leach SA. Effect of climate change on vector-borne disease risk in the UK. Lancet Infect Dis. 2015;15(6):721–30. doi:10.1016/S1473-3099(15)70091-5.
  • Tourapi C, Tsioutis C. Circular policy: a new approach to vector and vector-borne diseases’ management in line with the global vector control response (2017–2030). Trop Med Infect Dis. 2022;7(7):125. doi:10.3390/tropicalmed7070125.
  • Wilson AL, Courtenay O, Kelly-Hope LA, Scott TW, Takken W, Torr SJ, Lindsay SW. The importance of vector control for the control and elimination of vector-borne diseases. PLoS Negl Trop Dis. 2020;14(1):e0007831. doi:10.1371/journal.pntd.0007831.
  • Keiser J, Singer BH, Utzinger J. Reducing the burden of malaria in different eco-epidemiological settings with environmental management: a systematic review. Lancet Infect Dis. 2005;5(11):695–708. doi:10.1016/S1473-3099(05)70268-1.
  • Dahmana H, Mediannikov O. Mosquito-borne diseases emergence/resurgence and how to effectively control it biologically. Pathogens. 2020;9(4):310. doi:10.3390/pathogens9040310.
  • Hassan AO, Oso OV, Obeagu EI, Adeyemo AT. Malaria vaccine: prospects and challenges. Madonna Univ J Med Health Sci. 2022;2:22–40.
  • Coutinho-Abreu IV, Ramalho-Ortigao, Ramalho-Ortigao M, M. Transmission blocking vaccines to control insect-borne diseases: a review. Memórias do Inst Oswaldo Cruz. 2010;105(1):1–12. doi:10.1590/S0074-02762010000100001.
  • Laurens MB. RTS, S/AS01 vaccine (Mosquirix™): an overview. Hum Vaccines Immunother. 2020;16:480–9. doi:10.1080/21645515.2019.1669415.
  • Owino EA. World’s first malaria vaccine and its significance to malaria control in Africa. Asian Pac J Trop Med. 2022;15(2):49–52. doi:10.4103/1995-7645.338435.
  • Tully D, Griffiths CL. Dengvaxia: the world’s first vaccine for prevention of secondary dengue. Ther Adv Vaccines Immunother. 2021;9:25151355211015839. doi:10.1177/25151355211015839.
  • Salje H, Alera MT, Chua MN, Hunsawong T, Ellison D, Srikiatkhachorn A, Jarman RG, Gromowski GD, Rodriguez-Barraquer I, Cauchemez S, et al. Evaluation of the extended efficacy of the Dengvaxia vaccine against symptomatic and subclinical dengue infection. Nature Medicine. 2021;27:1395–400. doi:10.1038/s41591-021-01392-9.
  • Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71. doi:10.1136/bmj.n71.
  • Rivera L, Biswal S, Sáez-Llorens X, Reynales H, López-Medina E, Borja-Tabora C, Bravo L, Sirivichayakul C, Kosalaraksa P, Martinez Vargas L, et al. Three-year efficacy and safety of takeda’s dengue vaccine candidate (TAK-003). Clin Infect Dis. 2022;75(1):107–17. doi:10.1093/cid/ciab864.
  • Kallas EG, Precioso AR, Palacios R, Thomé B, Braga PE, Vanni T, Campos LMA, Ferrari L, Mondini G, da Graça Salomão M, et al. Safety and immunogenicity of the tetravalent, live-attenuated dengue vaccine Butantan-DV in adults in Brazil: a two-step, double-blind, randomised placebo-controlled phase 2 trial. Lancet Infect Dis. 2020;20(7):839–50. doi:10.1016/S1473-3099(20)30023-2.
  • Sridhar S, Luedtke A, Langevin E, Zhu M, Bonaparte M, Machabert T, Savarino S, Zambrano B, Moureau A, Khromava A, et al. Effect of dengue serostatus on dengue vaccine safety and efficacy. N Engl J Med. 2018;379(4):327–40. doi:10.1056/NEJMoa1800820.
  • Wichmann O, Vannice K, Asturias EJ, de Albuquerque Luna EJ, Longini I, Lopez AL, Smith PG, Tissera H, Yoon I-K, Hombach J, et al. Live-attenuated tetravalent dengue vaccines: the needs and challenges of post-licensure evaluation of vaccine safety and effectiveness. Vaccine. 2017;35(42):5535–42. doi:10.1016/j.vaccine.2017.08.066.
  • Sirivichayakul C, Barranco-Santana EA, Esquilin-Rivera I, Oh HM, Raanan M, Sariol CA, Shek LP, Simasathien S, Smith MK, Velez ID, et al. Safety and immunogenicity of a tetravalent dengue vaccine candidate in healthy children and adults in dengue-endemic regions: a randomized, placebo-controlled phase 2 Study. J Infect Dis. 2016;213(10):1562–72. doi:10.1093/infdis/jiv762.
  • Guy B, Briand O, Lang J, Saville M, Jackson N. Development of the Sanofi Pasteur tetravalent dengue vaccine: one more step forward. Vaccine. 2015;33(50):7100–11. doi:10.1016/j.vaccine.2015.09.108.
  • Villar L, Dayan GH, Arredondo-García JL, Rivera DM, Cunha R, Deseda C, Reynales H, Costa MS, Morales-Ramírez JO, Carrasquilla G, et al. Efficacy of a tetravalent dengue vaccine in children in Latin America. N Engl J Med. 2015;372(2):113–23. doi:10.1056/NEJMoa1411037.
  • Capeding MR, Tran NH, Hadinegoro SR, Ismail HI, Chotpitayasunondh T, Chua MN, Luong CQ, Rusmil K, Wirawan DN, Nallusamy R, et al. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. Lancet. 2014;384(9951):1358–65. doi:10.1016/S0140-6736(14)61060-6.
  • Dayan GH, Garbes P, Noriega F, Izoton de Sadovsky AD, Rodrigues PM, Giuberti C, Dietze R. Immunogenicity and safety of a recombinant tetravalent dengue vaccine in children and adolescents ages 9–16 years in Brazil. Am J Trop Med Hyg. 2013;89(6):1058–65. doi:10.4269/ajtmh.13-0304.
  • Durbin AP, Kirkpatrick BD, Pierce KK, Elwood D, Larsson CJ, Lindow JC, Tibery C, Sabundayo BP, Shaffer D, Talaat KR, et al. A single dose of any of four different live attenuated tetravalent dengue vaccines is safe and immunogenic in flavivirus-naive adults: a randomized, double-blind clinical trial. J Infect Dis. 2013;207(6):957–65. doi:10.1093/infdis/jis936.
  • Villar L, Rivera-Medina DM, Arredondo-García JL, Boaz M, Starr-Spires L, Thakur M, Zambrano B, Miranda MC, Rivas E, Dayan GH. Safety and immunogenicity of a recombinant tetravalent dengue vaccine in 9–16 year olds. Pediatr Infect Dis J. 2013;32(10):1102–9. doi:10.1097/INF.0b013e31829b8022.
  • Hss AS, Koh MT, Tan KK, Chan LG, Zhou L, Bouckenooghe A, Crevat D, Hutagalung Y. Safety and immunogenicity of a tetravalent dengue vaccine in healthy children aged 2–11 years in Malaysia: a randomized, placebo-controlled, phase III study. Vaccine. 2013;31(49):5814–21. doi:10.1016/j.vaccine.2013.10.013.
  • Lanata CF, Andrade T, Gil AI, Terrones C, Valladolid O, Zambrano B, Saville M, Crevat D. Immunogenicity and safety of tetravalent dengue vaccine in 2–11 year-olds previously vaccinated against yellow fever: randomized, controlled, phase II study in Piura, Peru. Vaccine. 2012;30(41):5935–41. doi:10.1016/j.vaccine.2012.07.043.
  • Sabchareon A, Wallace D, Sirivichayakul C, Limkittikul K, Chanthavanich P, Suvannadabba S, Jiwariyavej V, Dulyachai W, Pengsaa K, Wartel TA, et al. Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. Lancet. 2012;380(9853):1559–67. doi:10.1016/S0140-6736(12)61428-7.
  • Tran N, Luong C, Vu T, Forrat R, Lang J, Vu Q, Bouckenooghe A, Wartel TA. Safety and immunogenicity of recombinant, live attenuated tetravalent dengue vaccine (CYD-TDV) in healthy Vietnamese adults and children. J Vaccines Vaccin. 2012;3:1–8.
  • Osorio JE, Huang CY, Kinney RM, Stinchcomb DT. Development of DENVax: a chimeric dengue-2 PDK-53-based tetravalent vaccine for protection against dengue fever. Vaccine. 2011;29(42):7251–60. doi:10.1016/j.vaccine.2011.07.020.
  • Datoo MS, Natama HM, Somé A, Bellamy D, Traoré O, Rouamba T, Tahita MC, Ido NFA, Yameogo P, Valia D, et al. Efficacy and immunogenicity of R21/Matrix-M vaccine against clinical malaria after 2 years’ follow-up in children in Burkina Faso: a phase 1/2b randomised controlled trial. Lancet Infect Dis. 2022;22(12):1728–36. doi:10.1016/S1473-3099(22)00442-X.
  • Datoo MS, Natama MH, Somé A, Traoré O, Rouamba T, Bellamy D, Yameogo P, Valia D, Tegneri M, Ouedraogo F, et al. Efficacy of a low-dose candidate malaria vaccine, R21 in adjuvant matrix-M, with seasonal administration to children in Burkina Faso: a randomised controlled trial. Lancet. 2021;397(10287):1809–18. doi:10.1016/S0140-6736(21)00943-0.
  • Sirima SB, Mordmüller B, Milligan P, Ngoa UA, Kironde F, Atuguba F, Tiono AB, Issifou S, Kaddumukasa M, Bangre O, et al. A phase 2b randomized, controlled trial of the efficacy of the GMZ2 malaria vaccine in African children. Vaccine. 2016;34(38):4536–42. doi:10.1016/j.vaccine.2016.07.041.
  • Neafsey DE, Juraska M, Bedford T, Benkeser D, Valim C, Griggs A, Lievens M, Abdulla S, Adjei S, Agbenyega T, et al. Genetic diversity and protective efficacy of the RTS, S/AS01 malaria vaccine. N Engl J Med. 2015;373(21):2025–37. doi:10.1056/NEJMoa1505819.
  • RTS, S Clinical Trials Partnership. Efficacy, Safety of RTS, S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386(9988):31–45.
  • The Rts SCTP. Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLOS Med. 2014;11(7):e1001685. doi:10.1371/journal.pmed.1001685.
  • Moorthy VS, Imoukhuede EB, Milligan P, Bojang K, Keating S, Kaye P, Pinder M, Gilbert SC, Walraven G, Greenwood BM, et al. A randomised, double-blind, controlled vaccine efficacy trial of DNA/MVA ME-TRAP against malaria infection in Gambian adults. PLOS Med. 2004;1(2):e33. doi:10.1371/journal.pmed.0010033.
  • Olotu A, Fegan G, Wambua J, Nyangweso G, Awuondo KO, Leach A, Lievens M, Leboulleux D, Njuguna P, Peshu N, et al. Four-year efficacy of RTS,S/AS01E and its interaction with malaria exposure. N Engl J Med. 2013;368(12):1111–20. doi:10.1056/NEJMoa1207564.
  • RTS, S Clinical Trials Partnership. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med. 2012;367(24):2284–95. doi:10.1056/NEJMoa1208394.
  • Olotu A, Lusingu J, Leach A, Lievens M, Vekemans J, Msham S, Lang T, Gould J, Dubois M-C, Jongert E, et al. Efficacy of RTS,S/AS01E malaria vaccine and exploratory analysis on anti-circumsporozoite antibody titres and protection in children aged 5–17 months in Kenya and Tanzania: a randomised controlled trial. Lancet Infect Dis. 2011;11(2):102–9. doi:10.1016/S1473-3099(10)70262-0.
  • Asante KP, Abdulla S, Agnandji S, Lyimo J, Vekemans J, Soulanoudjingar S, Owusu R, Shomari M, Leach A, Jongert E,et al. Safety and efficacy of the RTS,S/AS01E candidate malaria vaccine given with expanded-programme-on-immunisation vaccines: 19 month follow-up of a randomised, open-label, phase 2 trial. Lancet Infect Dis. 2011;11(10):741–9. doi:10.1016/S1473-3099(11)70100-1.
  • Leach A, Vekemans J, Lievens M, Ofori-Anyinam O, Cahill C, Owusu-Agyei S, Abdulla S, Macete E, Njuguna P, Savarese B, et al. Design of a phase III multicenter trial to evaluate the efficacy of the RTS,S/AS01 malaria vaccine in children across diverse transmission settings in Africa. Malar J. 2011;10(1):224. doi:10.1186/1475-2875-10-224.
  • Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes JF, Abossolo BP, Conzelmann C. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med. 2011;365:1863–75.
  • Guinovart C, Aponte JJ, Sacarlal J, Aide P, Leach A, Bassat Q, Macete E, Dobaño C, Lievens M, Loucq C, et al. Insights into long-lasting protection induced by RTS, S/AS02A malaria vaccine: further results from a phase IIb trial in Mozambican children. PLOS ONE. 2009;4(4):e5165. doi:10.1371/journal.pone.0005165.
  • Sacarlal J, Aide P, Aponte JJ, Renom M, Leach A, Mandomando I, Lievens M, Bassat Q, Lafuente S, Macete E, et al. Long-term safety and efficacy of the RTS, S/AS02A malaria vaccine in Mozambican children. J Infect Dis. 2009;200(3):329–36. doi:10.1086/600119.
  • Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, Moris P, Schwenk R, Nielsen R, Debebe Z, Pinelis E, et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS, S/AS01B and RTS, S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J Infect Dis. 2009;200(3):337–46. doi:10.1086/600120.
  • Bejon P, Lusingu J, Olotu A, Leach A, Lievens M, Vekemans J, Mshamu S, Lang T, Gould J, Dubois M-C, et al. Efficacy of RTS, S/AS01E vaccine against malaria in children 5 to 17 months of age. N Engl J Med. 2008;359(24):2521–32. doi:10.1056/NEJMoa0807381.
  • Abdulla S, Oberholzer R, Juma O, Kubhoja S, Machera F, Membi C, Omari S, Urassa A, Mshinda H, Jumanne A, et al. Safety and immunogenicity of RTS, S/AS02D malaria vaccine in infants. N Engl J Med. 2008;359(24):2533–44. doi:10.1056/NEJMoa0807773.
  • Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG Jr, Hall T, Wellde BT, White K, Sun P, Schwenk R, et al. A phase I/IIa safety, immunogenicity, and efficacy bridging randomized study of a two-dose regimen of liquid and lyophilized formulations of the candidate malaria vaccine RTS,S/AS02A in malaria-naïve adults. Vaccine. 2007;25(29):5359–66. doi:10.1016/j.vaccine.2007.05.005.
  • Aponte JJ, Aide P, Renom M, Mandomando I, Bassat Q, Sacarlal J, Manaca MN, Lafuente S, Barbosa A, Leach A, et al. Safety of the RTS, S/AS02D candidate malaria vaccine in infants living in a highly endemic area of Mozambique: a double blind randomised controlled phase I/IIb trial. Lancet. 2007;370(9598):1543–51. doi:10.1016/S0140-6736(07)61542-6.
  • Bojang KA, Milligan PJM, Pinder M, Vigneron L, Alloueche A, Kester KE, Ballou WR, Conway DJ, Reece WH, Gothard P, et al. Efficacy of RTS, S/AS02 malaria vaccine against Plasmodium falciparum infection in semi-immune adult men in the Gambia: a randomised trial. Lancet. 2001;358(9297):1927–34. doi:10.1016/S0140-6736(01)06957-4.
  • Camacho LA, de Aguiar SG, Freire Mda S, Leal Mda L, Do Nascimento JP, Iguchi T, Lozana JA, Farias RH. Reactogenicity of yellow fever vaccines in a randomized, placebo-controlled trial. Rev Saude Publica. 2005;39(3):413–20. doi:10.1590/S0034-89102005000300012.
  • Monath TP, Nichols R, Archambault WT, Moore L, Marchesani R, Tian J, Shope RE, Thomas N, Schrader R, Furby D, et al. Comparative safety and immunogenicity of two yellow fever 17D vaccines (ARILVAX and YF-VAX) in a phase III multicenter, double-blind clinical trial. Am J Trop Med Hyg. 2002;66(5):533–41. doi:10.4269/ajtmh.2002.66.533.
  • McGuinness LA, Higgins JPT. Risk-of-bias VISualization (robvis): an R package and Shiny web app for visualizing risk-of-bias assessments. Res Synth Methods. 2020;12(1):55–61. doi:10.1002/jrsm.1411.
  • Hartling L, Ospina M, Liang Y, Dryden DM, Hooton N, Seida JK, Klassen TP. Risk of bias versus quality assessment of randomised controlled trials: cross sectional study. BMJ. 2009;339(oct19 1):4012–4012. doi:10.1136/bmj.b4012.
  • Hartling L, Ospina M, Liang Y, Dryden DM, Hooton N, Seida JK, Klassen TP. Risk of bias versus quality assessment of randomised controlled trials: cross sectional study. BMJ. 2009;339(oct19 1):b4012. doi:10.1136/bmj.b4012.
  • Pathak VK, Mohan M. A notorious vector-borne disease: dengue fever, its evolution as public health threat. J Family Med Prim Care. 2019;8(10):3125. doi:10.4103/jfmpc.jfmpc_716_19.
  • Organization W. Global strategy for dengue prevention and control, 2012–2020 WHO, Geneva 2012; 2012.
  • López-Medina E, Biswal S, Saez-Llorens X, Borja-Tabora C, Bravo L, Sirivichayakul C, Vargas LM, Alera MT, Velásquez H, Reynales H, et al. Efficacy of a dengue vaccine candidate (TAK-003) in healthy children and adolescents 2 years after vaccination. J Infect Dis. 2020;225(9):1521–32. doi:10.1093/infdis/jiaa761.
  • White LJ, Young EF, Stoops MJ, Henein SR, Adams EC, Baric RS, de Silva AM. Defining levels of dengue virus serotype-specific neutralizing antibodies induced by a live attenuated tetravalent dengue vaccine (TAK-003). PLOS Negl Trop Dis. 2021;15(3):e0009258. doi:10.1371/journal.pntd.0009258.
  • Tricou V, Folschweiller N, Lloyd E, Rauscher M, Biswal S. Efficacy and safety of Takeda’s tetravalent dengue vaccine candidate (TAK-003) after 4.5 years of follow-up. In: 44th ICMM World Congress on Military Medicine. Vol. 11. 2022. p. 5–9.
  • Durbin AP. Historical discourse on the development of the live attenuated tetravalent dengue vaccine candidate TV003/TV005. Curr Opin Virol. 2020;43:79–87. doi:10.1016/j.coviro.2020.09.005.
  • Kirkpatrick BD, Durbin AP, Pierce KK, Carmolli MP, Tibery CM, Grier PL, Hynes N, Diehl SA, Elwood D, Jarvis AP, et al. Robust and balanced immune responses to all 4 dengue virus serotypes following administration of a single dose of a live attenuated tetravalent dengue vaccine to healthy, flavivirus-naive adults. J Infect Dis. 2015;212(5):702–10. doi:10.1093/infdis/jiv082.
  • Silveira CG, Magnani DM, Costa PR, Avelino-SilvaI V, Ricciardi MJ, Timenetsky M. Plasmablast expansion following the tetravalent, live-attenuated dengue vaccine Butantan-DV in DENV-Naïve and DENV-exposed individuals in a Brazilian cohort. Front Immunol. 2022;13:908398. doi:10.3389/fimmu.2022.908398.
  • Thomas SJ, Yoon IK. A review of Dengvaxia®: development to deployment. Hum Vaccin Immunother. 2019;15(10):2295–314. doi:10.1080/21645515.2019.1658503.
  • Guy B, Barrere B, Malinowski C, Saville M, Teyssou R, Lang J. From research to phase III: preclinical, industrial and clinical development of the Sanofi Pasteur tetravalent dengue vaccine. Vaccine. 2011;29(42):7229–41. doi:10.1016/j.vaccine.2011.06.094.
  • Guy B, Saville M, Lang J. Development of Sanofi Pasteur tetravalent dengue vaccine. Hum Vaccin. 2010;6(9):696–705. doi:10.4161/hv.6.9.12739.
  • Dayan GH, Langevin E, Forrat R, Zambrano B, Noriega F, Frago C, Bouckenooghe A, Machabert T, Savarino S, DiazGranados CA, et al. Efficacy after 1 and 2 doses of CYD-TDV in dengue endemic areas by dengue serostatus. Vaccine. 2020;38(41):6472–7. doi:10.1016/j.vaccine.2020.07.056.
  • Malisheni M, Khaiboullina SF, Rizvanov AA, Takah N, Murewanhema G, Bates M. Clinical efficacy, safety, and immunogenicity of a live attenuated tetravalent dengue vaccine (CYD-TDV) in children: a systematic review with meta-analysis. Front Immunol. 2017;8:863. doi:10.3389/fimmu.2017.00863.
  • Mizumoto K, Ejima K, Yamamoto T, Nishiura H. On the risk of severe dengue during secondary infection: a systematic review coupled with mathematical modeling. J Vector Borne Dis. 2014;51:153.
  • Endy TP, Yoon I-K, Mammen MP. Current topics in microbiology and immunology. In: Dengue virus. Vol. 338. 2010. p. 1–13.
  • Guy B, Jackson N. Dengue vaccine: hypotheses to understand CYD-TDV-induced protection. Nat Rev Microbiol. 2016;14(1):45–54. doi:10.1038/nrmicro.2015.2.
  • Tran NH, Chansinghakul D, Chong CY, Low CY, Shek LP, Luong CQ, Frago C, Wartel TA, Sun S, Skipetrova A, et al. Long-term immunogenicity and safety of tetravalent dengue vaccine (CYD-TDV) in healthy populations in Singapore and Vietnam: 4-year follow-up of randomized, controlled, phase II trials. Hum Vaccines Immunother. 2019;15(10):2315–27. doi:10.1080/21645515.2019.1578595.
  • Bonam SR, Rénia L, Tadepalli G, Bayry J, Kumar HMS. Plasmodium falciparum malaria vaccines and vaccine adjuvants. Nato Vaccines. 2021;9(10):1072. doi:10.3390/vaccines9101072.
  • Rougeron V, Boundenga L, Arnathau C, Durand P, Renaud F, Prugnolle F. A population genetic perspective on the origin, spread and adaptation of the human malaria agents Plasmodium falciparum and Plasmodium vivax. FEMS Microbiol Rev. 2022;46(1):fuab047. doi:10.1093/femsre/fuab047.
  • Snow RW. Global malaria eradication and the importance of Plasmodium falciparum epidemiology in Africa. BMC Med. 2015;13(1):23. doi:10.1186/s12916-014-0254-7.
  • Garcia LS. Malaria. Clin Lab Med. 2010;30(1):93–129. doi:10.1016/j.cll.2009.10.001.
  • Theisen M, Soe S, Brunstedt K, Follmann F, Bredmose L, Israelsen H, Madsen SM, Druilhe P. A Plasmodium falciparum GLURP–MSP3 chimeric protein; expression in Lactococcus lactis, immunogenicity and induction of biologically active antibodies. Vaccine. 2004;22(9–10):1188–98. doi:10.1016/j.vaccine.2003.09.017.
  • Oeuvray C, Theisen M, Rogier C, Trape JF, Jepsen S, Druilhe P, Petri WA. Cytophilic immunoglobulin responses to Plasmodium falciparum glutamate-rich protein are correlated with protection against clinical malaria in Dielmo, Senegal. Infect Immun. 2000;68(5):2617–20. doi:10.1128/IAI.68.5.2617-2620.2000.
  • Roussilhon C, Oeuvray C, Müller-Graf C, Tall A, Rogier C, Trape JF, Theisen M, Balde A, Pérignon J-L, Druilhe P, et al. Long-term clinical protection from falciparum malaria is strongly associated with IgG3 antibodies to merozoite surface protein 3. PLoS Med. 2007;4(11):e320. doi:10.1371/journal.pmed.0040320.
  • Jepsen MP, Jogdand PS, Singh SK, Esen M, Christiansen M, Issifou S, Hounkpatin AB, Ateba-Ngoa U, Kremsner PG, Dziegiel MH, et al. The malaria vaccine candidate GMZ2 elicits functional antibodies in individuals from malaria endemic and non-endemic areas. J Infect Dis. 2013;208(3):479–88. doi:10.1093/infdis/jit185.
  • Esen M, Kremsner PG, Schleucher R, Gässler M, Imoukhuede EB, Imbault N, Leroy O, Jepsen S, Knudsen BW, Schumm M, et al. Safety and immunogenicity of GMZ2 — a MSP3–GLURP fusion protein malaria vaccine candidate. Vaccine. 2009;27(49):6862–8. doi:10.1016/j.vaccine.2009.09.011.
  • Mordmüller B, Szywon K, Greutelaers B, Esen M, Mewono L, Treut C, Mürbeth RE, Chilengi R, Noor R, Kilama WL, et al. Safety and immunogenicity of the malaria vaccine candidate GMZ2 in malaria-exposed, adult individuals from Lambaréné, Gabon. Vaccine. 2010;28(41):6698–703. doi:10.1016/j.vaccine.2010.07.085.
  • Bélard S, Issifou S, Hounkpatin AB, Schaumburg F, Ngoa UA, Esen M, Fendel R, de Salazar PM, Mürbeth RE, Milligan P, et al. A randomized controlled phase Ib trial of the malaria vaccine candidate GMZ2 in African children. PLOS ONE. 2011;6(7):e22525. doi:10.1371/journal.pone.0022525.
  • Clyde DF, Most H, McCarthy VC, Vanderberg JP. Immunization of man against sporozoite-induced falciparum malaria. Am J Med Sci. 1973;266(3):169–77. doi:10.1097/00000441-197309000-00002.
  • Vreden SG, Verhave JP, Oettinger T, Sauerwein RW, Meuwissen JH. Phase I clinical trial of a recombinant malaria vaccine consisting of the circumsporozoite repeat region of Plasmodium falciparum coupled to hepatitis B surface antigen. Am J Trop Med Hyg. 1991;45(5):533–8. doi:10.4269/ajtmh.1991.45.533.
  • Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, Desmons P, Wellde BT, Garçon N, Krzych U, Marchand M, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med. 1997;336(2):86–91. doi:10.1056/NEJM199701093360202.
  • Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG, Hall T, Krzych U, Delchambre M, Voss G, Dowler M, et al. Efficacy of recombinant circumsporozoite protein vaccine regimens against experimental Plasmodium falciparum malaria. J Infect Dis. 2001;183(4):640–7. doi:10.1086/318534.
  • Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, Moris P, Schwenk R, Nielsen RA, Debebe Z, Pinelis E, et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J Infect Dis. 2009;200(3):337–46. doi:10.1086/600120.
  • Polhemus ME, Remich SA, Ogutu BR, Waitumbi JN, Otieno L, Apollo S, Cummings JF, Kester KE, Ockenhouse CF, Stewart A, et al. Evaluation of RTS, S/AS02A and RTS, S/AS01B adults in high malar transmission area. PLOS ONE. 2009;4(7):e6465. doi:10.1371/journal.pone.0006465.
  • Penny MA, Verity R, Bever CA, Sauboin C, Galactionova K, Flasche S, White MT, Wenger EA, Van de Velde N, Pemberton-Ross P, et al. Public health impact and cost-effectiveness of the RTS,S/AS01 malaria vaccine: a systematic comparison of predictions from four mathematical models. Lancet. 2016;387(10016):367–75. doi:10.1016/S0140-6736(15)00725-4.
  • Otieno L, Oneko M, Otieno W, Abuodha J, Owino E, Odero C, Mendoza YG, Andagalu B, Awino N, Ivinson K, et al. Safety and immunogenicity of RTS,S/AS01 malaria vaccine in infants and children with WHO stage 1 or 2 HIV disease: a randomised, double-blind, controlled trial. Lancet Infect Dis. 2016;16(10):1134–44. doi:10.1016/S1473-3099(16)30161-X.
  • Lell B, Agnandji S, von Glasenapp I, Haertle S, Oyakhiromen S, Issifou S, Vekemans J, Leach A, Lievens M, Dubois M-C, et al. A randomized trial assessing the safety and immunogenicity of AS01 and AS02 adjuvanted RTS,S malaria vaccine candidates in children in Gabon. PLOS ONE. 2009;4(10):e7611. doi:10.1371/journal.pone.0007611.
  • Owusu-Agyei S, Ansong D, Asante K, Kwarteng Owusu S, Owusu R, Wireko Brobby NA, Dosoo D, Osei Akoto A, Osei-Kwakye K, Adjei EA, et al. Randomized controlled trial of RTS,S/AS02D and RTS,S/AS01E malaria candidate vaccines given according to different schedules in Ghanaian children. PLOS ONE. 2009;4(10):e7302. doi:10.1371/journal.pone.0007302.
  • Pallikkuth S, Chaudhury S, Lu P, Pan L, Jongert E, Wille-Reece U, Pahwa S. A delayed fractionated dose RTS, S AS01 vaccine regimen mediates protection via improved T follicular helper and B cell responses. Elife. 2020;9:e51889. doi:10.7554/eLife.51889.
  • Garçon N, Di Pasquale A. From discovery to licensure, the Adjuvant System story. Hum Vaccines Immunother. 2017;13(1):19–33. doi:10.1080/21645515.2016.1225635.
  • Regules JA, Cummings JF, Ockenhouse CF. The RTS, S vaccine candidate for malaria. Expert Rev Vaccines. 2011;10(5):589–99. doi:10.1586/erv.11.57.
  • Stoute J, Kester K, Krzych U, Wellde B, Hall T, White K, Glenn G, Ockenhouse CF, Garcon N, Schwenk R, et al. Long-term efficacy and immune responses following immunization with the RTS, S malaria vaccine. J Infect Dis. 1998;178(4):1139–44. doi:10.1086/515657.
  • Leroux-Roels G, Leroux-Roels I, Clement F, Ofori-Anyinam O, Lievens M, Jongert E, Moris P, Ballou WR, Cohen J. Evaluation of the immune response to RTS,S/AS01 and RTS,S/AS02 adjuvanted vaccines: randomized, double-blind study in malaria-naïve adults. Hum Vaccin Immunother. 2014;10(8):2211–9. doi:10.4161/hv.29375.
  • Garçon N, Heppner DG, Cohen J. Development of RTS,S/AS02: a purified subunit-based malaria vaccine candidate formulated with a novel adjuvant. Expert Rev Vaccines. 2003;2:231–8. doi:10.1586/14760584.2.2.231.
  • Collins KA, Snaith R, Cottingham MG, Gilbert SC, Hill AVS. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci Rep. 2017;7(1):46621. doi:10.1038/srep46621.
  • Venkatraman N, Tiono AB, Bowyer G, Powlson J, Collins KA, Coulibaly S, Datoo M, Silman D, Ouedraogo A, Nébié I, et al. Phase I assessments of first-in-human administration of a novel malaria anti-sporozoite vaccine candidate, R21 in matrix-M adjuvant, in UK and Burkinabe volunteers. medRxiv. 2019:19009282.
  • Krishna S. Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLOS Med. 2014;11(7):e1001685. doi:10.1371/journal.pmed.1001685.
  • Geneva. WHO recommends R21/Matrix-M vaccine for malaria prevention in updated advice on immunization; 2023.
  • Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801. doi:10.1016/j.cell.2006.02.015.
  • Ogwang C, Kimani D, Edwards NJ, Roberts R, Mwacharo J, Bowyer G, Bliss C, Hodgson SH, Njuguna P, Viebig NK, et al. Prime-boost vaccination with chimpanzee adenovirus and modified vaccinia Ankara encoding TRAP provides partial protection against Plasmodium falciparum infection in Kenyan adults. Sci Transl Med. 2015;7(286):2865–5. doi:10.1126/scitranslmed.aaa2373.
  • Afolabi MO, Tiono AB, Adetifa UJ, Yaro JB, Drammeh A, Nébié I, Bliss C, Hodgson SH, Anagnostou NA, Sanou GS, et al. Safety and immunogenicity of ChAd63 and MVA ME-TRAP in West African children and infants. Mol Ther. 2016;24(8):1470–7. doi:10.1038/mt.2016.83.
  • Bejon P, Mwacharo J, Kai O, Todryk S, Keating S, Lowe B, Lang T, Mwangi TW, Gilbert SC, Peshu N, et al. The induction and persistence of T cell IFN-γ responses after vaccination or natural exposure is suppressed by Plasmodium falciparum. J Immunol. 2007;179(6):4193–201. doi:10.4049/jimmunol.179.6.4193.